KR102329524B1 - Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient - Google Patents

Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient Download PDF

Info

Publication number
KR102329524B1
KR102329524B1 KR1020210103567A KR20210103567A KR102329524B1 KR 102329524 B1 KR102329524 B1 KR 102329524B1 KR 1020210103567 A KR1020210103567 A KR 1020210103567A KR 20210103567 A KR20210103567 A KR 20210103567A KR 102329524 B1 KR102329524 B1 KR 102329524B1
Authority
KR
South Korea
Prior art keywords
let7
liver cancer
modified
composition
present
Prior art date
Application number
KR1020210103567A
Other languages
Korean (ko)
Inventor
남석우
양희두
Original Assignee
주식회사 네오나
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 주식회사 네오나 filed Critical 주식회사 네오나
Priority to KR1020210103567A priority Critical patent/KR102329524B1/en
Priority to PCT/KR2021/014873 priority patent/WO2023013818A1/en
Application granted granted Critical
Publication of KR102329524B1 publication Critical patent/KR102329524B1/en
Priority to AU2022324176A priority patent/AU2022324176A1/en
Priority to PCT/KR2022/011190 priority patent/WO2023013990A1/en
Priority to CA3228077A priority patent/CA3228077A1/en

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/14Type of nucleic acid interfering N.A.
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification

Abstract

The present invention relates to a pharmaceutical composition for preventing or treating liver cancer, comprising modified RT-LET7 as an active ingredient. The modified RT-LET7 enhances the suppression of the expression of Let-7i-5p compared with existing RT-LET7, enhances the suppression of the growth of liver cancer cells, increases TSP1 expression, and enhances macrophage phagocytic activity. Therefore, the modified RT-LET7 can be used as a pharmaceutical composition for preventing or treating liver cancer.

Description

변형된 RT-LET7을 유효성분으로 포함하는 간암의 예방 또는 치료용 조성물{COMPOSITION FOR PREVENTING OR TREATING OF LIVER CANCER COMPRISING MODIFIED RT-LET7 AS AN ACTIVE INGREDIENT}A composition for preventing or treating liver cancer comprising a modified RT-LET7 as an active ingredient {COMPOSITION FOR PREVENTING OR TREATING OF LIVER CANCER COMPRISING MODIFIED RT-LET7 AS AN ACTIVE INGREDIENT}

본 발명은 간암의 예방 또는 치료용 약학 조성물에 관한 것으로, 구체적으로, Let-7i-5p의 발현 억제제인 변형된 RT-LET7을 유효성분으로 포함하는 간암의 예방 또는 치료용 약학 조성물에 관한 것이다.The present invention relates to a pharmaceutical composition for preventing or treating liver cancer, and more particularly, to a pharmaceutical composition for preventing or treating liver cancer comprising a modified RT-LET7, an expression inhibitor of Let-7i-5p, as an active ingredient.

간암은 전 세계적으로 다섯 번째로 빈발하는 암이지만 그로 인한 사망률은 3위에 해당하는 공격적인 암이다(Ahn J, Flamm SL Hepatocellular carcinoma Dis Mon 2004;50:556-573) 치료목적의 수술은 단지 15%에서 25% 정도의 환자에게만 가능하고 대부분의 간암 환자들은 국부적으로 진행하거나 전이되는 질병들에 의해 비교적 단기간 내에 사망한다(Roberts LR, Gores GJ Hepatocellular carcinoma: molecular pathways and new therapeutic targets Semin Liver Dis 2005;25:212-225) 간암의 주요 원인으로 B형 간염 바이러스(hepatitis B virus), C형 간염 바이러스(hepatitis C virus), 및 아플라톡신 B1(aflatoxin B1) 등이 잘 알려져 있다. 하지만, 지난 20년간 간암 환자의 전체적인 생존율은 크게 증가하지 않았고, 간암의 발달(development) 및 진전(progression) 기작은 여전히 잘 알려져 있지 않은 상태이다(Bruix J, et al Focus on hepatocellular carcinoma Cancer Cell 2004;5:215-219) 지금까지, 분자표적치료(molecular targeted therapy)가 성숙한 간암의 치료에 효과적인 것으로 나타났지만(Shen YC, Hsu C, Cheng AL Molecular targeted therapy for advanced hepatocellular carcinoma: current status and future perspectives J Gastroenterol;45:794-807), 어떻게 이러한 유전적 변화가 간암 환자들 개개인에게 관찰되는 임상적 특징들을 야기하는지는 불명확하다.Although liver cancer is the fifth most common cancer worldwide, it is the third most aggressive cancer in mortality (Ahn J, Flamm SL Hepatocellular carcinoma Dis Mon 2004;50:556-573). It is possible in only 25% of patients, and most liver cancer patients die within a relatively short period of time due to locally advanced or metastatic diseases (Roberts LR, Gores GJ Hepatocellular carcinoma: molecular pathways and new therapeutic targets Semin Liver Dis 2005;25: 212-225) Hepatitis B virus, hepatitis C virus, and aflatoxin B1 are well known as major causes of liver cancer. However, the overall survival rate of liver cancer patients has not increased significantly over the past 20 years, and the mechanism of development and progression of liver cancer is still unknown (Bruix J, et al Focus on hepatocellular carcinoma Cancer Cell 2004; 5:215-219) So far, molecular targeted therapy has been shown to be effective for the treatment of mature liver cancer (Shen YC, Hsu C, Cheng AL Molecular targeted therapy for advanced hepatocellular carcinoma: current status and future perspectives J Gastroenterol;45:794-807), it is unclear how these genetic changes lead to the clinical features observed in individual HCC patients.

HDACs (Histone deacetylases)는 종종 보조억제자(corepressors)나 다중-단백질 전사복합체(multi-protein transcriptional complexes)들에 의해 유전자 프로모터에 붙을 수 있으며, 그곳에서 DNA에 직접 결합하지 않고 크로마틴(chromatin) 변형을 통해 전사를 조절한다(Thiagalingam S, Cheng KH, Lee HJ, Mineva N, Thiagalingam A, Ponte JF Histone deacetylases: unique players in shaping the epigenetic histone code Ann N Y Acad Sci 2003;983:84-100) 암호화된 사람 HDACs는 18개가 있으며, 이들은 클래스 I (HDAC 1, 2, 3 및 8), 클래스 II (HDAC 4, 5, 6, 7, 9 및 10), 클래스 III (SIRT 1-7), 및 클래스 IV (HDAC11) 효소들로 분류된다(Yang XJ, Seto E The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men Nat Rev Mol Cell Biol 2008;9:206-218) 히스톤 아세틸화 효소(acetyltransferases) 및 HDACs 모두 세포 증식, 분화 및 세포주기 조절에 관여한다는 사실이 알려져 있다(Witt O, Deubzer HE, Milde T, Oehme I HDAC family: What are the cancer relevant targets Cancer Lett 2009;277:8-21) 또한, HDACs의 병리학적 활성 및 조절감소(deregulation)가 암, 면역질환, 및 근이영양증(muscular dystrophy)과 같은 여러 질병들을 야기할 수 있다는 사실이 보고되었다(Yang XJ, Seto E HATs and HDACs: from structure, function and regulation to novel strategies for therapy and prevention Oncogene 2007;26:5310-5318). HDAC6는 HDACs의 클래스 IIb 패밀리 멤버이고, 미세소관(MTs)[0004] 과 관련 있는 세포질내 탈아세틸화효소(cytoplasmic deacetylase)로 작용하며, 알파-튜뷸린(α-tubulin)을 탈아세틸화시킨다(Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, et al HDAC6 is a microtubule-associated Mis18α. Nature 2002;417:455-458). Histone deacetylases (HDACs) can often be attached to gene promoters by corepressors or multi-protein transcriptional complexes, where they modify chromatin without binding directly to DNA. (Thiagalingam S, Cheng KH, Lee HJ, Mineva N, Thiagalingam A, Ponte JF Histone deacetylases: unique players in shaping the epigenetic histone code Ann NY Acad Sci 2003;983:84-100) There are 18 HDACs, which are class I (HDAC 1, 2, 3 and 8), class II (HDAC 4, 5, 6, 7, 9 and 10), class III (SIRT 1-7), and class IV ( HDAC11) enzymes (Yang XJ, Seto E The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men Nat Rev Mol Cell Biol 2008;9:206-218) Histone acetyltransferases and HDACs are known to be involved in cell proliferation, differentiation and cell cycle regulation (Witt O, Deubzer HE, Milde T, Oehme I HDAC family: What are the cancer relevant targets Cancer Lett 2009;277:8-21) , it has been reported that pathological activity and deregulation of HDACs can cause several diseases such as cancer, immune disease, and muscular dystrophy (Yang XJ, Seto E HATs and HDACs: from structure, functi on and regulation to novel strategies for therapy and prevention Oncogene 2007;26:5310-5318). HDAC6 is a class IIb family member of HDACs, acts as a cytoplasmic deacetylase related to microtubules (MTs), and deacetylates α-tubulin ( Hubbert C, Guardiola A, Shao R, Kawaguchi Y, Ito A, Nixon A, et al HDAC6 is a microtubule-associated Mis18α. Nature 2002;417:455-458).

한편, miRNA는 세포 내에 존재하는 20-25 핵산(nucleotide) 길이의 작은 RNA (endogenous small RNA)의 일종으로 단백질을 합성하지 않는 DNA에서 유래되어 헤어핀-구조 전사체(hairpin-shaped transcript)로부터 생성이 된다. miRNA는 표적 mRNA의 3'-UTR의 상보적인 서열에 결합하여 그 mRNA의 번역 억제 또는 불안정화를 유도하여, 궁극적으로 그 표적 mRNA의 단백질 합성을 억제하는 리프레서(repressor) 역할을 하게 된다. 하나의 miRNA는 여러 개의 mRNA를 타겟팅하며, mRNA 역시 여러 개의 miRNA에 의해 조절될 수 있다고 알려져 있다.On the other hand, miRNA is a type of endogenous small RNA with a length of 20-25 nucleotides present in cells. do. miRNA binds to the complementary sequence of the 3'-UTR of the target mRNA, induces translational inhibition or destabilization of the mRNA, and ultimately serves as a repressor to inhibit protein synthesis of the target mRNA. One miRNA targets multiple mRNAs, and it is known that mRNA can also be regulated by multiple miRNAs.

한편, 대식세포들은 식균작용에 의해 질병세포(암세포)들을 탐식하는데, 그것은 항체들의 Fc 절편이 대식세포들 막표면의 Fc 수용체와의 결합을 매개함으로써 일어난다. 그러나 종양들은 정상 면역 조절 기구 전복을 통해 대식세포들을 포함한 면역세포들의 공격에서 벗어날 수 있다. 그러한 기전의 하나가 정상 세포들에서 발현되는 하나의 단백질인 CD47이다. CD47은 SIRPα(signal-regulatory protein α)라 부르는 대식세포의 수용체와 상호작용하여 이것이 대식세포들에게 “나를 잡아먹지 말라(탐식 차단)”는 신호 전달을 이끌음으로써 정상 세포들을 떠나게 한다. 동일하게 암세포들에 의한 CD47 발현도 그들로 하여금 암세포들이 항체와 결합했을 경우에서 조차 대식세포들에 대한 저항성을 갖게 하는 것이다. 따라서 종양들에 많은 수의 대식세포들이 접근하지만 “탐식 차단” 신호가 꺼지지 않으면 암세포들에게 작용할 수없는 것이다. 이에대한 하나의 치료 전략이 CD47에 대한 단일클론 항체를 이용한 “탐식 차단” 신호를 차단하는 것이다.On the other hand, macrophages phagocytose diseased cells (cancer cells) by phagocytosis, which occurs because Fc fragments of antibodies mediate binding to Fc receptors on the membrane surface of macrophages. However, tumors can escape from attack by immune cells, including macrophages, by overthrowing the normal immune regulatory machinery. One such mechanism is CD47, a protein expressed in normal cells. CD47 interacts with a macrophage receptor called SIRPα (signal-regulatory protein α), which leads to the macrophage “do not eat me (phagocytic blockade)” signal, which causes normal cells to leave. Equally, CD47 expression by cancer cells makes them resistant to macrophages even when the cancer cells bind to antibodies. Thus, although a large number of macrophages access the tumors, they cannot act on cancer cells unless the “phagocytic block” signal is turned off. One treatment strategy for this is to block the “phagocytic blocking” signal using a monoclonal antibody against CD47.

따라서, 본 발명은 Let-7i-5p의 발현 억제제인 RT-LET7을 변형시켜 기존의 RT-LET7 보다 효과가 상승됨을 통해 이를 간암의 예방 또는 치료용 약학 조성물 로 사용될 수 있음을 확인하였고, 또한 변형된 RT-LET7에 의한 대식세포 식작용을 조절할 수 있음을 통해 CD47-양성 간암의 치료용 면역항암제 약학 조성물로 사용될 수 있음을 확인하여 본 발명을 완성하였다.Therefore, the present invention confirmed that it can be used as a pharmaceutical composition for the prevention or treatment of liver cancer by modifying RT-LET7, an expression inhibitor of Let-7i-5p, and increasing the effect than the existing RT-LET7, and also modified By regulating macrophage phagocytosis by RT-LET7, it was confirmed that it can be used as an immunotherapy pharmaceutical composition for the treatment of CD47-positive liver cancer, thereby completing the present invention.

KRUS 10-2012-001489310-2012-0014893 AA

본 발명의 목적은 간암의 예방 또는 치료용 약학 조성물을 제공하는 것이다.It is an object of the present invention to provide a pharmaceutical composition for preventing or treating liver cancer.

또한, 본 발명의 목적은 CD47-양성 간암의 치료용 면역항암제 약학 조성물을 제공하는 것이다.Another object of the present invention is to provide a pharmaceutical composition for immunotherapy for the treatment of CD47-positive liver cancer.

상기 과제를 해결하기 위하여, 본 발명은 변형된 RT-LET7을 유효성분으로 포함하는 간암의 예방 또는 치료용 약학 조성물을 제공한다.In order to solve the above problems, the present invention provides a pharmaceutical composition for preventing or treating liver cancer comprising a modified RT-LET7 as an active ingredient.

또한, 본 발명은 변형된 RT-LET7을 유효성분으로 포함하는 CD47-양성 간암의 치료용 면역항암제 약학 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for immunotherapy for the treatment of CD47-positive liver cancer comprising modified RT-LET7 as an active ingredient.

본 발명에 따른 변형된 RT-LET7은 기존 간암세포의 Let-7i-5p를 억제하는 안티센스 microRNA(AS-miRNA)인 RT-LET7 대비 간암에서의 종양 세포 성장을 현저하게 억제하는 것을 확인할 수 있었고, 변형된 RT-LET7은 간암세포에서 Let-7i-5p를 억제해 TSP1을 증가시키고, 증가한 TSP1은 CD47과 결합해 대식세포의 SIRPα와 상호작용을 막게되며, 따라서 간암세포의 CD47과 대식세포의 SIRPα 결합을 막아 암세포에 대한 대식세포의 면역활성을 높여 항암효과를 보이는 기전을 통해 대식세포 식작용 활성을 현저히 증가시킴을 확인하여, 이를 간암의 예방 및 치료용 약학 조성물 또는 CD47-양성 간암의 예방 또는 치료용 면역항암제 약학 조성물로 이용할 수 있음을 확인하였다.It was confirmed that the modified RT-LET7 according to the present invention significantly inhibited tumor cell growth in liver cancer compared to RT-LET7, which is an antisense microRNA (AS-miRNA) that inhibits Let-7i-5p of existing liver cancer cells, Modified RT-LET7 increases TSP1 by inhibiting Let-7i-5p in hepatocarcinoma cells, and the increased TSP1 binds to CD47 and blocks the interaction with SIRPα of macrophages. Therefore, CD47 of hepatocarcinoma cells and SIRPα of macrophages By blocking binding, it was confirmed that macrophage phagocytic activity was significantly increased through the mechanism of showing anticancer effect by increasing the immune activity of macrophages against cancer cells, and this was confirmed as a pharmaceutical composition for the prevention and treatment of liver cancer or CD47-positive liver cancer prevention or treatment It was confirmed that it can be used as a pharmaceutical composition for immunotherapy.

도 1은 Let-7i-5p의 발현 억제제인 RT-LET7의 다양한 변형 구조(도 1a) 및 억제 효율(도 1b)을 나타낸 도이다(기본형: RT-LET7, 변형: RT-LET7-2, RT-LET7-4, RT-LET7-6 및 RT-LET7-8).
도 2는 본 발명의 변형된 RT-LET7인 RT-LET7-8이 HCC 세포주에서 유의미하게 지속적으로 Let-7i-5p를 억제하는 효과를 확인한 도이다(도 2a; SNU-387, 도 2b; SNU-368, 도 2c; SNU-423).
도 3은 본 발명의 변형된 RT-LET7인 RT-LET7-8에 의해 HCC 세포주의 생장억제 및 대식세포의 활성 증가를 확인한 도이다(도 3a; MTT 및 세포 생존 분석을 통한 Let-7i-5p의 종양 형성에 대한 특성, 도 3b; 기본형 RT-LET7 및 변형 RT-LET7-8을 처리 후 웨스턴 블롯 분석을 통해 TSP1 발현변화, 도 3c; 기본형 RT-LET7 및 변형 RT-LET7-8가 처리된 HCC 세포의 대식세포 식작용 활성).
1 is a diagram showing various modified structures (FIG. 1A) and inhibition efficiency (FIG. 1B) of RT-LET7, an expression inhibitor of Let-7i-5p (basic type: RT-LET7, modification: RT-LET7-2, RT -LET7-4, RT-LET7-6 and RT-LET7-8).
2 is a diagram confirming the effect of RT-LET7-8, a modified RT-LET7 of the present invention, to significantly and continuously inhibit Let-7i-5p in HCC cell lines (FIG. 2a; SNU-387, FIG. 2b; SNU); -368, Fig. 2c; SNU-423).
3 is a view confirming the growth inhibition of HCC cell line and increase in macrophage activity by RT-LET7-8, which is a modified RT-LET7 of the present invention (FIG. 3a; Let-7i-5p through MTT and cell survival analysis) Characteristics of tumorigenesis, Figure 3b; TSP1 expression change through western blot analysis after treatment with primitive RT-LET7 and modified RT-LET7-8, Figure 3c; Basic type RT-LET7 and modified RT-LET7-8 treated macrophage phagocytosis of HCC cells).

이하, 첨부된 도면을 참조하여 본 발명의 구현예로 본 발명을 상세히 설명하기로 한다. 다만, 하기 구현예는 본 발명에 대한 예시로 제시되는 것으로, 당업자에게 주지 저명한 기술 또는 구성에 대한 구체적인 설명이 본 발명의 요지를 불필요하게 흐릴 수 있다고 판단되는 경우에는 그 상세한 설명을 생략할 수 있고, 이에 의해 본 발명이 제한되지는 않는다. 본 발명은 후술하는 특허청구범위의 기재 및 그로부터 해석되는 균등 범주 내에서 다양한 변형 및 응용이 가능하다. Hereinafter, the present invention will be described in detail as an embodiment of the present invention with reference to the accompanying drawings. However, the following embodiments are presented as examples of the present invention, and when it is determined that detailed descriptions of well-known techniques or configurations known to those skilled in the art may unnecessarily obscure the gist of the present invention, the detailed description may be omitted, and , the present invention is not limited thereby. Various modifications and applications of the present invention are possible within the scope of equivalents interpreted therefrom and the description of the claims to be described later.

또한, 본 명세서에서 사용되는 용어(terminology)들은 본 발명의 바람직한 실시예를 적절히 표현하기 위해 사용된 용어들로서, 이는 사용자, 운용자의 의도 또는 본 발명이 속하는 분야의 관례 등에 따라 달라질 수 있다. 따라서, 본 용어들에 대한 정의는 본 명세서 전반에 걸친 내용을 토대로 내려져야 할 것이다. 명세서 전체에서, 어떤 부분이 어떤 구성요소를 "포함"한다고 할 때, 이는 특별히 반대되는 기재가 없는 한 다른 구성요소를 제외하는 것이 아니라 다른 구성 요소를 더 포함할 수 있는 것을 의미한다.In addition, the terms used in this specification are terms used to properly express the preferred embodiment of the present invention, which may vary according to the intention of a user or operator, or customs in the field to which the present invention belongs. Accordingly, definitions of these terms should be made based on the content throughout this specification. Throughout the specification, when a part "includes" a certain component, it means that other components may be further included, rather than excluding other components, unless otherwise stated.

일 측면에서, 본 발명은 서열번호 1로 표시되는 핵산분자로서, Let-7i-5p를 표적화하는 핵산분자에 관한 것이다.In one aspect, the present invention relates to a nucleic acid molecule represented by SEQ ID NO: 1, which targets Let-7i-5p.

일 구현예에서, 본 발명의 "핵산분자"는 서열번호 1의 뉴클레오티드 서열이 2`-O-methyl(2`-OMe)화 또는 2`-O-methoxyethyl(2`-O-MOE)화 되어 변형된 것일 수 있다.In one embodiment, in the "nucleic acid molecule" of the present invention, the nucleotide sequence of SEQ ID NO: 1 is 2`-O-methyl (2`-OMe) or 2`-O-methoxyethyl (2`-O-MOE) may be deformed.

일 구현예에서, 본 발명의 "핵산분자"는 서열번호 1의 뉴클레오티드 서열의 3`을 Phosphorothioate로 변형된 것일 수 있다.In one embodiment, the "nucleic acid molecule" of the present invention may be one in which 3' of the nucleotide sequence of SEQ ID NO: 1 is modified with Phosphorothioate.

일 구현예에서, 본 발명의 "핵산분자"는 서열번호 1의 뉴클레오티드 서열의 5`말단으로부터 4번째까지 서열의 뉴클레오티드 중 어느하나 이상의 뉴클레오티드 서열의 3`이 Phosphorothioate로 변형된 것일 수 있다.In one embodiment, the "nucleic acid molecule" of the present invention may be one in which 3' of any one or more nucleotide sequences of the nucleotide sequence from the 5' end to the 4th of the nucleotide sequence of SEQ ID NO: 1 is modified with phosphorothioate.

일 구현예에서, 본 발명의 "핵산분자"는 서열번호 1의 뉴클레오티드 서열의 3`말단으로부터 4번째까지 서열의 뉴클레오티드 중 어느하나 이상의 뉴클레오티드 서열의 3`이 Phosphorothioate로 변형된 것일 수 있다.In one embodiment, the "nucleic acid molecule" of the present invention may be one in which 3' of any one or more of the nucleotide sequences of the nucleotide sequence from the 3' end to the 4th of the nucleotide sequence of SEQ ID NO: 1 is modified with phosphorothioate.

일 구현예에서, 본 발명의 "핵산분자"는 서열번호 1의 뉴클레오티드 서열의 5`말단으로부터 4번째까지 서열의 뉴클레오티드 중 어느 하나 이상의 뉴클레오티드 서열의 3`이 Phosphorothioate로 변형된 것이며, 서열번호 1의 뉴클레오티드 서열의 3`말단으로부터 4번째까지 서열의 뉴클레오티드 중 어느 하나 이상의 뉴클레오티드 서열의 3`이 Phosphorothioate로 변형된 것일 수 있다.In one embodiment, the "nucleic acid molecule" of the present invention is one in which 3' of any one or more nucleotide sequences of the nucleotide sequence from the 5' end to the 4th of the nucleotide sequence of SEQ ID NO: 1 is modified with Phosphorothioate, 3' of any one or more nucleotide sequences among the nucleotides from the 3' end to the 4th of the nucleotide sequence may be modified with phosphorothioate.

일 구현예에서, 본 발명의 "핵산분자"는 서열번호 1의 뉴클레오티드 서열의 5`말단으로부터 4번째까지 서열의 뉴클레오티드 모든 뉴클레오티드의 3`이 Phosphorothioate로 변형된 것이며, 서열번호 1의 뉴클레오티드 서열의 3`말단으로부터 4번째까지 서열의 모든 뉴클레오티드 서열의 3`이 Phosphorothioate로 변형된 것일 수 있다.In one embodiment, the "nucleic acid molecule" of the present invention is one in which 3' of all nucleotides of the nucleotide sequence from the 5' end to the 4th of the nucleotide sequence of SEQ ID NO: 1 are modified with Phosphorothioate, and 3 of the nucleotide sequence of SEQ ID NO: 1 '3 of all nucleotide sequences from the end to the 4th' may be modified with Phosphorothioate.

일 측면에서, 본 발명은 상기 핵산분자를 유효성분으로 포함하는 간암의 예방 또는 치료용 약학 조성물에 관한 것이다.In one aspect, the present invention relates to a pharmaceutical composition for preventing or treating liver cancer comprising the nucleic acid molecule as an active ingredient.

일 구현예에서, 본 발명의 "조성물"은 Let-7i-5p의 발현을 억제하는 것일 수 있다.In one embodiment, the "composition" of the present invention may inhibit the expression of Let-7i-5p.

일 구현예에서, 본 발명의 "조성물"은 간암 세포 성장을 억제하는 것일 수 있다.In one embodiment, the "composition" of the present invention may inhibit liver cancer cell growth.

일 구현예에서, 본 발명의 "조성물"은 TSP1(thrombospondin-1) 발현을 증가시키는 것일 수 있다.In one embodiment, the "composition" of the present invention may increase TSP1 (thrombospondin-1) expression.

일 구현예에서, 본 발명의 "조성물"은 대식세포 식작용 활성을 증가시키는 것일 수 있다.In one embodiment, the "composition" of the present invention may increase macrophage phagocytosis activity.

일 측면에서, 본 발명은 상기 핵산분자를 유효성분으로 포함하는 CD47-양성 간암의 치료용 면역항암제 약학 조성물에 관한 것이다.In one aspect, the present invention relates to an immunotherapy pharmaceutical composition for the treatment of CD47-positive liver cancer comprising the nucleic acid molecule as an active ingredient.

일 구현예에서, 본 발명의 "TSP1"은 CD47 수용체를 점유하여 CD47-SIRPα의 상호작용을 방해할 수 있다.In one embodiment, "TSP1" of the present invention is capable of interfering with CD47-SIRPα interaction by occupying the CD47 receptor.

일 구현예에서, 본 발명의 "핵산분자"는 let-7i-p-TSP1 신호전달 축을 조절할 수 있으며, 대식세포와 HCC 사이의 CD47-SIRPα 상호작용을 CD47-TSP1 상호작용으로 전환함으로써 대식세포의 HCC 세포에 대한 식작용을 재-활성화하는 것일 수 있다.In one embodiment, the "nucleic acid molecule" of the present invention can modulate the let-7i-p-TSP1 signaling axis, and converts the CD47-SIRPα interaction between macrophages and HCC into a CD47-TSP1 interaction in macrophages. re-activating phagocytosis on HCC cells.

일 구현예에서, 본 발명의 "간암"은 Let-7i-5p 고발현 간암일 수 있으며, 간세포암종(hepatocellular carcinoma)일 수 있고, stage Ⅰ, Ⅱ, Ⅲ, ⅣA 또는 ⅣB 병기(phase)의 간세포암종일 수 있으며, 초기 병기보다 치료가 어려운 stageⅢ 내지 Ⅳ 병기의 간세포암종인 것이 더욱 바람직하다.In one embodiment, the "liver cancer" of the present invention may be Let-7i-5p high-expressing liver cancer, may be hepatocellular carcinoma, and stage I, II, III, IVA or IVB stage hepatocellular carcinoma. It may be a carcinoma, and more preferably stage III to IV hepatocellular carcinoma, which is more difficult to treat than the initial stage.

일 구현예에서, 본 발명의 "간암"은 TSP1 저발현 간암일 수 있으며, 간세포암종일 수 있고, stage Ⅰ, Ⅱ, Ⅲ, ⅣA 또는 ⅣB 병기(phase)의 간세포암종일 수 있으며, 초기 병기보다 치료가 어려운 stageⅢ 내지 Ⅳ 병기의 간세포암종인 것이 더욱 바람직하다.In one embodiment, the "liver cancer" of the present invention may be TSP1 low-expressing liver cancer, may be hepatocellular carcinoma, may be hepatocellular carcinoma of stage I, II, III, IVA or IVB, and may be at an earlier stage. It is more preferable that it is stage III to IV hepatocellular carcinoma, which is difficult to treat.

본 발명에서 사용되는 용어, "발현 억제"란 표적 유전자의 (mRNA로의) 발현 또는 (단백질로의) 번역 저하를 야기하는 것을 의미하며, 바람직하게는 이에 의해 표적 유전자 발현이 탐지 불가능해지거나 무의미한 수준으로 존재하게 되는 것을 의미한다.As used herein, the term “repression of expression” means to cause a decrease in expression (to mRNA) or translation (to protein) of a target gene, preferably, whereby the expression of the target gene becomes undetectable or insignificant. means to exist as

본 발명에서, 사용된 용어 "예방"이란 본 발명에 따른 조성물의 투여에 의해 간암의 발생, 발달 및 재발을 억제 또는 지연시키는 모든 행위를 의미한다.In the present invention, the term "prevention" as used means any action of inhibiting or delaying the occurrence, development and recurrence of liver cancer by administration of the composition according to the present invention.

본 발명에서 사용된 용어 "치료"란 본 발명에 따른 조성물의 투여로 간암 및 이로 인한 합병증의 증세를 호전시키거나 이롭게 변경하는 모든 행위를 의미한다. 본 발명이 속하는 기술분야에서 통상의 지식을 가진 자라면, 대한의학협회 등에서 제시된 자료를 참조하여 본원의 조성물이 효과가 있는 질환의 정확한 기준을 알고, 개선, 향상 및 치료된 정도를 판단할 수 있을 것이다.As used herein, the term “treatment” refers to any act of improving or beneficially changing the symptoms of liver cancer and its complications by administering the composition according to the present invention. Those of ordinary skill in the art to which the present invention pertains, with reference to the data presented in the Korean Medical Association, etc., know the exact criteria of the disease for which the composition of the present application is effective, and can determine the degree of improvement, improvement and treatment will be.

본 발명의 조성물의 치료적으로 유효한 양은 여러 요소, 예를 들면 투여방법, 목적부위, 개체의 상태 등에 따라 달라질 수 있다. The therapeutically effective amount of the composition of the present invention may vary depending on several factors, for example, the administration method, the target site, the condition of the subject, and the like.

본 발명의 약학적조성물은 약학적으로 유효한 양으로 투여한다. 본 발명에서 사용되는 용어, "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미하며, 유효 용량 수준은 개체의 건강상태, 감염증의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 본 발명의 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와 순차적으로 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 상기한 요소들을 모두 고려하여, 부작용없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.The pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount. As used herein, the term "pharmaceutically effective amount" means an amount sufficient to treat a disease at a reasonable benefit/risk ratio applicable to medical treatment and not to cause side effects, and the effective dose level is Health status, type and severity of infection, drug activity, sensitivity to drug, administration method, administration time, administration route and excretion rate, treatment period, factors including drugs used in combination or concurrently, and other factors well known in the medical field can be determined according to The composition of the present invention may be administered as an individual therapeutic agent or in combination with other therapeutic agents, may be administered sequentially or simultaneously with conventional therapeutic agents, and may be administered singly or multiple times. In consideration of all of the above factors, it is important to administer an amount that can obtain the maximum effect with a minimum amount without side effects, which can be easily determined by those skilled in the art.

일 구현예에서, 상기 약학 조성물은 경구형 제형, 외용제, 좌제, 멸균 주사용액 및 분무제를 포함하는 군으로부터 선택되는 하나 이상의 제형일 수 있다. In one embodiment, the pharmaceutical composition may be one or more formulations selected from the group comprising oral formulations, topical formulations, suppositories, sterile injection solutions and sprays.

본 발명의 조성물은 또한 생물학적 제제에 통상적으로 사용되는 담체, 희석제, 부형제 또는 둘 이상의 이들의 조합을 포함할 수 있다. 약학적으로 허용 가능한 담체는 조성물을 생체 내 전달에 적합한 것이면 특별히 제한되지 않으며, 예를 들면, Merck Index, 13th ed., Merck & Co. Inc. 에 기재된 화합물, 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로스 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 이용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한, 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주이용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. 더 나아가 당 분야의 적정한 방법으로 또는 Remington's Pharmaceutical Science(Mack Publishing Company, Easton PA, 18th, 1990)에 개시되어 있는 방법을 이용하여 각 질환에 따라 또는 성분에 따라 바람직하게 제제화할 수 있다.The compositions of the present invention may also include carriers, diluents, excipients or combinations of two or more commonly used in biological agents. The pharmaceutically acceptable carrier is not particularly limited as long as it is suitable for in vivo delivery of the composition, for example, Merck Index, 13th ed., Merck & Co. Inc. Compounds described in , saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol, and one or more of these components can be mixed and used, and if necessary, other antioxidants, buffers, bacteriostats, etc. Conventional additives may be added. In addition, diluents, dispersants, surfactants, binders and lubricants may be additionally added to formulate into injectable formulations such as aqueous solutions, suspensions, emulsions, pills, capsules, granules or tablets. Furthermore, it can be preferably formulated according to each disease or component using an appropriate method in the art or a method disclosed in Remington's Pharmaceutical Science (Mack Publishing Company, Easton PA, 18th, 1990).

본 발명의 조성물에 추가로 동일 또는 유사한 기능을 나타내는 유효성분을 1종 이상 함유할 수 있다. 본 발명의 조성물은, 조성물 총 중량에 대하여 상기 단백질을 0.0001 내지 10 중량 %로, 바람직하게는 0.001 내지 1 중량 %를 포함한다. The composition of the present invention may further contain one or more active ingredients exhibiting the same or similar function. The composition of the present invention comprises 0.0001 to 10% by weight of the protein, preferably 0.001 to 1% by weight, based on the total weight of the composition.

본 발명의 약학 조성물은 약학적으로 허용 가능한 첨가제를 더 포함할 수 있으며, 이때 약학적으로 허용 가능한 첨가제로는 전분, 젤라틴화 전분, 미결정셀룰로오스, 유당, 포비돈, 콜로이달실리콘디옥사이드, 인산수소칼슘, 락토스, 만니톨, 엿, 아라비아고무, 전호화전분, 옥수수전분, 분말셀룰로오스, 히드록시프로필셀룰로오스, 오파드라이, 전분글리콜산나트륨, 카르나우바 납, 합성규산알루미늄, 스테아린산, 스테아린산마그네슘, 스테아린산알루미늄, 스테아린산칼슘, 백당, 덱스트로스, 소르비톨 및 탈크 등이 사용될 수 있다. 본 발명에 따른 약학적으로 허용 가능한 첨가제는 상기 조성물에 대해 0.1 중량부 내지 90 중량부 포함되는 것이 바람직하나, 이에 한정되는 것은 아니다.The pharmaceutical composition of the present invention may further include a pharmaceutically acceptable additive, wherein the pharmaceutically acceptable additive includes starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, Lactose, mannitol, syrup, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, Opadry, sodium starch glycolate, lead carnauba, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, stearic acid Calcium, sucrose, dextrose, sorbitol, talc and the like can be used. The pharmaceutically acceptable additive according to the present invention is preferably included in an amount of 0.1 to 90 parts by weight based on the composition, but is not limited thereto.

본 발명의 조성물은 목적하는 방법에 따라 비 경구 투여(예를 들어 정맥 내, 피하, 복강 내 또는 국소에 적용)하거나 경구 투여할 수 있으며, 경구 투여 하는 것이 가장 바람직하다. 투여량은 개체의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 투여방법, 배설률 및 질환의 중증도 등에 따라 그 범위가 다양하다. The composition of the present invention may be administered parenterally (eg, intravenously, subcutaneously, intraperitoneally or topically) or orally according to a desired method, and oral administration is most preferred. The dosage varies according to the subject's body weight, age, sex, health status, diet, administration time, administration method, excretion rate, and severity of disease.

본 발명의 조성물의 경구 투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데, 통상적으로 사용되는 단순 희석제인 물, 액체 파라핀 이외에 다양한 부형제, 예컨대 습윤제, 감미제, 방향제, 보존제 등이 함께 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성 용제, 현탁제, 유제, 동결건조 제제, 좌제 등이 포함된다.Liquid preparations for oral administration of the composition of the present invention include suspensions, internal solutions, emulsions, syrups, etc., and various excipients, such as wetting agents, sweeteners, fragrances, and preservatives, in addition to commonly used simple diluents such as water and liquid paraffin. and the like may be included. Formulations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, freeze-dried preparations, suppositories, and the like.

하기의 실시예를 통하여 본 발명을 보다 상세하게 설명한다. 그러나 하기 실시예는 본 발명의 내용을 구체화하기 위한 것일 뿐 이에 의해 본 발명이 한정되는 것은 아니다.The present invention will be described in more detail through the following examples. However, the following examples are only for specifying the contents of the present invention, and the present invention is not limited thereto.

<실시예 1> 기존 RT-LET7의 변형된 RT-LET7 제작<Example 1> Manufacture of modified RT-LET7 of existing RT-LET7

Let-7i-5p의 억제제인 RT-LET7(서열번호1: AACAGCACAAACUACUACCUCA에 4단계 과정으로 한 cycle씩 진행하여, RNA oligo 1mer 씩 3‘말단에서 5’말단까지 Deblocking -> Coupling -> Oxidation -> Capping의 과정으로 순차적으로 진행한다. 이때, 변형된 염기를 사용하는데, Nat Rev Drug Discov. 2020 Oct;19(10):673-694.에 따르면, 2’-Ribose의 변형(2’-O-methyl, 2‘-MOE)은 변형시키지 않은 RNA보다 핵산분해효소로부터 저항성을 증가시키고, 세포질내에서 안정성을 증가시키는데 기여한다. 또한, 생체 조직내에서 반감기를 증가시키는 역할도 하여, 결과적으로 약물의 효과를 증대시키는 역할을 한다. 더욱이, 상보적인 RNA에 강하게 부착하여, RNase에 의해 표적 유전자가 더 효과적으로 분해되는데 도움을 준다. 추가로, RNA 염기사이에 phosphorothioate로 치환하였을 때, RNase의 활성에는 영향을 주지 않으며, 표적 RNA에 효율적으로 결합한다. 그리고, 세포와 체내에서 알부민과 같은 단백질과 결합하여, 핵산분해효소로부터 보호되고, 이에 따라, 약물이 소변으로 배출되는 것을 막아주는 역할을 하여, 체내에서 약물의 지속시간을 늘려, 약물의 효과를 증대시키는 역할을 하는 이유 때문에 Ribose와 phosphorothioate 변형을 진행하였다. 변형된 RNA 염기 2‘-OMe rA Phosphoramidite(Glen Research, Sterling, VA, cat. 10-3100), 2‘-OMe rC Phosphoramidite(Glen Research, cat. 10-3115), 2‘-OMe rG Phosphoramidite(Glen Research, cat. 10-3120), 2‘-OMe rU Phosphoramidite(Glen Research, cat. 10-3130), 2'-MOE rA Phosphoramidite(Glen Research, cat. 10-3200), 2'-MOE rC Phosphoramidite(Glen Research, cat. 10-3211), 2'-MOE rG Phosphoramidite(Glen Research, cat. 10-3220), 2'-MOE rU Phosphoramidite(Glen Research, cat. 10-3231)를 합성한다. 합성과정에서 phosphorothioate로 치환이 필요한 부분은 Sulfurizing Reagent II(Glen Research, cat. 40-4037-10)를 활용하여 합성한다(바이오니아 합성). 이때, 아무변형을 하지 않은 RNA oligo를 RT-LET7, 모든 염기를 2’-O-methyl로 변형하고, phosphorothioate로 변형한 RNA oligo를 RT-LET7-2, 5’말단과 3’말단 4곳을 변형시킨 염기를 RT-LET7-6으로 명명하였다. 그리고, 모든 염기를 2’-MOE로 변형하고, phosphorothioate로 변형한 RNA oligo를 RT-LET7-4, 5’말단과 3’말단 4곳을 변형시킨 염기를 RT-LET7-8로 명명하였다(도 1a).RT-LET7, an inhibitor of Let-7i-5p (SEQ ID NO: 1: AACAGCACAAACUACUCUCCUCA), proceeds one cycle in a four-step process, deblocking each 1mer of RNA oligo from the 3' end to the 5' end -> Coupling -> Oxidation -> Capping At this time, a modified base is used, and according to Nat Rev Drug Discov. 2020 Oct;19(10):673-694., the modification of 2'-Ribose (2'-O-methyl , 2'-MOE) increases resistance from nucleases compared to unmodified RNA, and contributes to increasing stability in the cytoplasm.In addition, it also serves to increase half-life in living tissues, resulting in drug effects Moreover, it strongly attaches to complementary RNA, helping target gene to be more effectively degraded by RNase In addition, when phosphorothioate is substituted between RNA bases, the activity of RNase is not affected. It binds efficiently to the target RNA and binds to proteins such as albumin in cells and in the body, protecting it from nucleases, and thus preventing the drug from being excreted in the urine, Ribose and phosphorothioate were modified for the reason that it increases the duration of the drug and enhances the effect of the drug.The modified RNA base 2'-OMe rA Phosphoramidite (Glen Research, Sterling, VA, cat. 10-3100) , 2'-OMe rC Phosphoramidite (Glen Research, cat. 10-3115), 2'-OMe rG Phosphoramidite (Glen Research, cat. 10-3120), 2'-OMe rU Phosphoramidite (Glen Research, cat. 10-3130) ), 2'-MOE rA Phosphoramidite (G len Research, cat. 10-3200), 2'-MOE rC Phosphoramidite (Glen Research, cat. 10-3211), 2'-MOE rG Phosphoramidite (Glen Research, cat. 10-3220), 2'-MOE rU Phosphoramidite (Glen Research, cat. 10-3231) is synthesized. The part that needs to be replaced with phosphorothioate in the synthesis process is synthesized using Sulfurizing Reagent II (Glen Research, cat. 40-4037-10) (Bioneer synthesis). At this time, the unmodified RNA oligo was RT-LET7, all bases were modified with 2'-O-methyl, and the RNA oligo modified with phosphorothioate was RT-LET7-2, 4 places at the 5' and 3' ends. The modified base was designated as RT-LET7-6. Then, all bases were modified with 2'-MOE, and the RNA oligo modified with phosphorothioate was named RT-LET7-4, and the bases modified at the 5' and 3' ends were named RT-LET7-8 (Fig. 1a).

<실시예 2> 변형된 RT-LET7의 처리에 의한 Let-7i-5p의 발현 억제 확인<Example 2> Confirmation of inhibition of Let-7i-5p expression by treatment of modified RT-LET7

도 1a와 같이 만들어진 변형된 RT-LET7를 트랜스펙션한 HCC 세포주(SNU-387, SNU-368, 및 SNU-423)에서 총 RNA를 TRIzol 시약(Invitrogen, Carlsbad, CA)을 이용하여 분리한 후, 상기 두 miRNA에 특이적인 cDNA를 micscipt II RT 키트(Qiagen, Manchester, UK)를 이용하여 합성하였다. qRT-PCR은 SensiFASTTM SYBR NoROX Kit(Bioline, London, UK)로 수행되었다. HCC 세포주(SNU-387, SNU-368, 및 SNU-423)에서 Let-7i-5p의 qRT-PCR 분석을 수행한 결과, 2’-MOE로 모든 염기를 치환 하였을 때(RT-LET7-4 및 RT-LET7-8), 2‘-O-methyl로 치환하였을 때(RT-LET7-2 및 RT-LET7-6) 보다 Let-7i-5p의 발현을 억제하는데 효과가 좋은 것을 확인하였다(도 1b).After isolation of total RNA from HCC cell lines (SNU-387, SNU-368, and SNU-423) transfected with the modified RT-LET7 made as shown in FIG. 1A using TRIzol reagent (Invitrogen, Carlsbad, CA) , cDNA specific for the two miRNAs was synthesized using the micscipt II RT kit (Qiagen, Manchester, UK). qRT-PCR was performed with a SensiFAST™ SYBR NoROX Kit (Bioline, London, UK). As a result of qRT-PCR analysis of Let-7i-5p in HCC cell lines (SNU-387, SNU-368, and SNU-423), when all bases were substituted with 2'-MOE (RT-LET7-4 and RT-LET7-8), it was confirmed that when substituted with 2'-O-methyl (RT-LET7-2 and RT-LET7-6), it was more effective in inhibiting the expression of Let-7i-5p (Fig. 1b). ).

또한, 2’-MOE로 RT-LET7을 변형시켰을 때, HCC 세포주(SNU-387, SNU-368, 및 SNU-423)에 트랜스펙션 후 최대 7일까지 배양 후 RNA 추출 후 Let-7i-5p의 발현 억제정도를 비교하였을 때, 2’-MOE로 변형시킨 후 phosphorothioate로 치환하였을 때, 아무 변형도 안한 RT-LET7에 비해서 Let-7i-5p의 발현 억제가 유의미하게 효율이 증대되는 것을 확인하였다(도 2).In addition, when RT-LET7 was modified with 2'-MOE, HCC cell lines (SNU-387, SNU-368, and SNU-423) were cultured for up to 7 days after transfection and RNA extraction after Let-7i-5p When comparing the degree of inhibition of the expression of Let-7i-5p, it was confirmed that the efficiency of expression inhibition of Let-7i-5p was significantly increased when modified with 2'-MOE and substituted with phosphorothioate compared to RT-LET7 without any modification. (Fig. 2).

<실시예 3> MTT 및 세포 생존 분석을 통한 Let-7i-5p의 종양 형성에 대한 특성 확인<Example 3> Characterization of Let-7i-5p for tumorigenesis through MTT and cell viability assay

간암에서의 RT-LET7 처리시 인비트로 종양형성(in vitro tumorigenesis) 억제능을 확인하기 위해, RT-LET7를 이용하여 MTT 분석을 수행하였다. 구체적으로, MTT 분석을 위해, SNU-387 세포를 12-웰 플레이트에 분주하고 RT-LET7, RT-LET7-4, RT-LET7-8을 트랜스펙션한 뒤, MTT[3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] 용액(Sigma) 0.5 mg/ml과 1시간 동안 인큐베이션한 뒤, SYNERGY H1 Multilabel 플레이트 리더기(Bio-Tek, Winooski, VT)를 이용하여 흡광도를 측정하였다. 그 결과, RT-LET7의 변형된 RT-LET7-8을 처리한 실험군에서 종양 세포 성장을 가장 우수하게 억제하는 것을 확인할 수 있었다(도 3a).In order to confirm the inhibitory ability of in vitro tumorigenesis upon RT-LET7 treatment in liver cancer, MTT analysis was performed using RT-LET7. Specifically, for MTT analysis, SNU-387 cells were aliquoted in a 12-well plate and transfected with RT-LET7, RT-LET7-4, RT-LET7-8, and then MTT[3-(4,5) After incubation with 0.5 mg/ml of -dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] solution (Sigma) for 1 hour, absorbance was measured using a SYNERGY H1 Multilabel plate reader (Bio-Tek, Winooski, VT). measured. As a result, it was confirmed that the most excellent inhibition of tumor cell growth in the experimental group treated with RT-LET7-8 modified RT-LET7 (FIG. 3a).

<실시예 4> 변형된 RT-LET7에 의한 대식세포 식작용 조절 확인<Example 4> Confirmation of macrophage phagocytosis regulation by modified RT-LET7

Let-7i-5p가 대식세포 식작용(macrophage phagocytosis)의 조절에 관여하는 TSP1의 발현을 조절하는지 확인하기 위해, RT-LET7과 변형된 RT-LET7-8을 처리 후 웨스턴 블롯 분석을 통해 발현변화를 확인하였다. 그 결과, RT-LET7과 RT-LET7-8을 각각 처리하였을 때, TSP1의 발현이 증가되는 것을 확인할 수 있었다(도 3b).To confirm whether Let-7i-5p regulates the expression of TSP1 involved in the regulation of macrophage phagocytosis, the expression change was analyzed by Western blot analysis after treatment with RT-LET7 and modified RT-LET7-8. Confirmed. As a result, when RT-LET7 and RT-LET7-8 were treated, it was confirmed that the expression of TSP1 was increased (FIG. 3b).

이러한 결과를 토대로, Let-7i-5p-TSP1 네트워크의 CD47과 SIRPα(signal regulatory protein α)의 상호작용에서 TSP1이 CD47 수용체를 점유하여 CD47-SIRPα의 상호작용을 방해하여 대식세포가 HCC를 식작용할 수 있게 하는지를 확인하기 위하여, 인비트로 식작용 분석을 수행하였다. 구체적으로, HCC 세포주인 SNU-387 각각 단일 세포 현탁액으로 만든 후, CFSE(abcam, Cambridge, UK)로 표지하였다. 그 후, C57BL/6 마우스로부터 복막 대식세포를 수득하고 SNU-387과 2시간 동안 공배양한 뒤, RT-LET7 및 변형된 RT-LET7-8로 각각 처리하였고, 양성 대조군으로 TSP1 재조합 단백질을 직접 처리한 HCC 세포와 비교하였다. 식작용 지수는 종양 세포를 포획하는 대식세포의 수를 대식세포의 총 수로 나눠 계산하였다. 그 결과, RT-LET7 및 변형된 RT-LET7-8가 처리된 HCC 세포 모두 대식세포 식작용 활성이 현저히 증가함을 확인하였고, RT-LET7 대비 변형된 RT-LET7-8에서 대식세포 식작용 활성이 더 증가함을 확인하였다(도 3c).Based on these results, in the interaction between CD47 and SIRPα (signal regulatory protein α) in the Let-7i-5p-TSP1 network, TSP1 occupies the CD47 receptor and interferes with the CD47-SIRPα interaction, so macrophages can phagocytic HCC. To confirm whether it is possible, an in vitro phagocytosis assay was performed. Specifically, each of the HCC cell lines, SNU-387, was prepared as a single cell suspension and labeled with CFSE (abcam, Cambridge, UK). Thereafter, peritoneal macrophages were obtained from C57BL/6 mice and co-cultured with SNU-387 for 2 hours, treated with RT-LET7 and modified RT-LET7-8, respectively, and TSP1 recombinant protein was directly treated as a positive control. compared to treated HCC cells. The phagocytosis index was calculated by dividing the number of macrophages capturing tumor cells by the total number of macrophages. As a result, both RT-LET7 and modified RT-LET7-8-treated HCC cells confirmed that macrophage phagocytosis activity was significantly increased, and macrophage phagocytosis activity was higher in modified RT-LET7-8 compared to RT-LET7. It was confirmed that the increase (Fig. 3c).

이를 통해, RT-LET7을 변형시킨 RT-LET7-8가 let-7i-p-TSP1 신호전달 축을 조절할 수 있으며, 대식세포와 HCC 사이의 CD47-SIRPα 상호작용을 CD47-TSP1 상호작용으로 전환함으로써 대식세포의 HCC 세포에 대한 식작용을 재-활성화하는 것을 알 수 있다.Through this, RT-LET7-8-modified RT-LET7 can regulate the let-7i-p-TSP1 signaling axis, and by converting the CD47-SIRPα interaction between macrophages and HCC into a CD47-TSP1 interaction, It can be seen that re-activation of phagocytosis on HCC cells of phagocytes.

<110> NEORNAT <120> COMPOSITION FOR PREVENTING OR TREATING OF LIVER CANCER COMPRISING MODIFIED RT-LET7 AS AN ACTIVE INGREDIENT <130> PN2106-323 <160> 1 <170> KoPatentIn 3.0 <210> 1 <211> 22 <212> RNA <213> Artificial Sequence <220> <223> RT-LET7 <400> 1 aacagcacaa acuacuaccu ca 22 <110> NEORAT <120> COMPOSITION FOR PREVENTING OR TREATING OF LIVER CANCER COMPRISING MODIFIED RT-LET7 AS AN ACTIVE INGREDIENT <130> PN2106-323 <160> 1 <170> KoPatentIn 3.0 <210> 1 <211> 22 <212> RNA <213> Artificial Sequence <220> <223> RT-LET7 <400> 1 aacagcacaa acuacuaccu ca 22

Claims (13)

서열번호 1의 뉴클레오티드 서열로 표시되는 Let-7i-5p를 표적화하는 핵산분자로서,
상기 핵산분자는,
서열번호 1의 뉴클레오티드의 모든 서열을 2’-O-methoxyethyl(2’-O-MOE)화 시켜 변형된 것이고,
서열번호 1의 뉴클레오티드 서열의 5’말단으로부터 4번째까지 뉴클레오티드의 3’이 Phosphorothioate로 변형된 것이며,
서열번호 1의 뉴클레오티드 서열의 3’말단으로부터 4번째까지 뉴클레오티드의 3’이 Phosphorothioate로 변형된 것인, Let-7i-5p를 표적화하는 핵산분자.
A nucleic acid molecule targeting Let-7i-5p represented by the nucleotide sequence of SEQ ID NO: 1,
The nucleic acid molecule is
All of the nucleotide sequences of SEQ ID NO: 1 are modified by 2'-O-methoxyethyl (2'-O-MOE),
3' of the nucleotide from the 5' end to the 4th of the nucleotide sequence of SEQ ID NO: 1 is modified with Phosphorothioate,
A nucleic acid molecule targeting Let-7i-5p, wherein the 3' of the nucleotide from the 3' to the 4th of the nucleotide sequence of SEQ ID NO: 1 is modified with Phosphorothioate.
삭제delete 삭제delete 삭제delete 삭제delete 삭제delete 삭제delete 제1항의 핵산분자를 유효성분으로 포함하는 간암의 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating liver cancer comprising the nucleic acid molecule of claim 1 as an active ingredient. 제8항에 있어서,
상기 조성물은 Let-7i-5p의 발현을 억제하는 것인, 간암의 예방 또는 치료용 약학 조성물.
9. The method of claim 8,
The composition inhibits the expression of Let-7i-5p, a pharmaceutical composition for preventing or treating liver cancer.
제8항에 있어서,
상기 조성물은 간암 세포 성장을 억제하는 것인, 간암의 예방 또는 치료용 약학 조성물.
9. The method of claim 8,
The composition inhibits the growth of liver cancer cells, a pharmaceutical composition for the prevention or treatment of liver cancer.
제8항에 있어서,
상기 조성물은 TSP1(thrombospondin-1) 발현을 증가시키는 것인, 간암의 예방 또는 치료용 약학 조성물.
9. The method of claim 8,
The composition is to increase TSP1 (thrombospondin-1) expression, a pharmaceutical composition for the prevention or treatment of liver cancer.
제8항에 있어서,
상기 조성물은 대식세포 식작용 활성을 증가시키는 것인, 간암의 예방 또는 치료용 약학 조성물.
9. The method of claim 8,
The composition is to increase macrophage phagocytosis activity, a pharmaceutical composition for the prevention or treatment of liver cancer.
제1항의 핵산분자를 유효성분으로 포함하는 CD47-양성 간암의 치료용 면역항암제 약학 조성물.An immunotherapy pharmaceutical composition for the treatment of CD47-positive liver cancer comprising the nucleic acid molecule of claim 1 as an active ingredient.
KR1020210103567A 2021-08-06 2021-08-06 Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient KR102329524B1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
KR1020210103567A KR102329524B1 (en) 2021-08-06 2021-08-06 Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient
PCT/KR2021/014873 WO2023013818A1 (en) 2021-08-06 2021-10-22 Composition for prevention or treatment of liver cancer comprising modified rt-let7 as active ingredient
AU2022324176A AU2022324176A1 (en) 2021-08-06 2022-07-29 Composition for prevention or treatment of liver cancer, comprising modified rt-let7 as active ingredient
PCT/KR2022/011190 WO2023013990A1 (en) 2021-08-06 2022-07-29 Composition for prevention or treatment of liver cancer, comprising modified rt-let7 as active ingredient
CA3228077A CA3228077A1 (en) 2021-08-06 2022-07-29 Composition for prevention or treatment of liver cancer, comprising modified rt-let7 as active ingredient

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
KR1020210103567A KR102329524B1 (en) 2021-08-06 2021-08-06 Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient

Publications (1)

Publication Number Publication Date
KR102329524B1 true KR102329524B1 (en) 2021-11-23

Family

ID=78695333

Family Applications (1)

Application Number Title Priority Date Filing Date
KR1020210103567A KR102329524B1 (en) 2021-08-06 2021-08-06 Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient

Country Status (1)

Country Link
KR (1) KR102329524B1 (en)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007112754A2 (en) * 2006-04-03 2007-10-11 Santaris Pharma A/S Pharmaceutical compositions comprising anti-mirna antisense oligonucleotides
KR20120014893A (en) 2009-04-08 2012-02-20 바스프 코포레이션 Zoned catalysts for diesel applications
WO2012119051A2 (en) * 2011-03-02 2012-09-07 Groove Biopharma Corporation Enhanced biodistribution of oligomers
KR20150006742A (en) * 2013-07-09 2015-01-19 (주)바이오니아 Liver cancer related genes-specific siRNA, double-stranded oligo RNA molecules comprising the siRNA, and composition for the prevention or treatment of cancer comprising the same
KR20150095349A (en) * 2014-02-13 2015-08-21 서울대학교산학협력단 Composition and method for treating neurodegenerative diseases targeting miRNA
KR20200119538A (en) * 2019-04-10 2020-10-20 가톨릭대학교 산학협력단 Composition for preventing or treating of liver cancer

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007112754A2 (en) * 2006-04-03 2007-10-11 Santaris Pharma A/S Pharmaceutical compositions comprising anti-mirna antisense oligonucleotides
KR20120014893A (en) 2009-04-08 2012-02-20 바스프 코포레이션 Zoned catalysts for diesel applications
WO2012119051A2 (en) * 2011-03-02 2012-09-07 Groove Biopharma Corporation Enhanced biodistribution of oligomers
KR20150006742A (en) * 2013-07-09 2015-01-19 (주)바이오니아 Liver cancer related genes-specific siRNA, double-stranded oligo RNA molecules comprising the siRNA, and composition for the prevention or treatment of cancer comprising the same
KR20150095349A (en) * 2014-02-13 2015-08-21 서울대학교산학협력단 Composition and method for treating neurodegenerative diseases targeting miRNA
KR20200119538A (en) * 2019-04-10 2020-10-20 가톨릭대학교 산학협력단 Composition for preventing or treating of liver cancer

Similar Documents

Publication Publication Date Title
EP2093292A2 (en) Inhibition of specific histone deacetylase isoforms
KR102344598B1 (en) Composition for preventing or treating of liver cancer
US20160168577A1 (en) Clusterin Antisense Therapy for Treatment of Cancer
AU2017232496B2 (en) Compositions and methods for the treatment of a beta-catenin-associated disease or disorder
WO2016068600A1 (en) Composition for treating cancer stem cells
JP2019533697A (en) 5-Halouracil modified microRNA and its use in the treatment of cancer
CN110201172B (en) Application of YY1 expression inhibitor in preparation of medicine for treating breast cancer
KR102329524B1 (en) Composition for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient
US20030152557A1 (en) Methods for inhibiting histone deacetylase-4
KR102428121B1 (en) Delivery system for preventing or treating of liver cancer comprising modified rt-let7 as an active ingredient
CN112274507A (en) Application of small-molecule inhibitor remodelin of NAT10 in preparation of oral squamous cell carcinoma treatment drug
CN108236722B (en) Application of IDNK inhibitor in preparation of liver cancer treatment drug
WO2023013818A1 (en) Composition for prevention or treatment of liver cancer comprising modified rt-let7 as active ingredient
KR20240017196A (en) Delivery system for preventing or treating of cancer comprising modified rt-let7 as an active ingredient
KR101855900B1 (en) Pharmaceutical composition for preventing or treating anticancer drug resistant breast cancers comprising expression or activity inhibitor of SETD1A as an active ingredient
WO2023013990A1 (en) Composition for prevention or treatment of liver cancer, comprising modified rt-let7 as active ingredient
KR102576855B1 (en) Composition for preventing or treating of liver cancer comprising tsp1 as an active ingredient
JP2020530844A (en) How to Treat Cancer by Inhibiting SETD2
KR101525229B1 (en) Pharmaceutical composition for the treatment of cancers or inhibition of cancer metastasis containing the inhibitors of Gpr171 expression or activity
KR101436684B1 (en) Composition for preventing or treating of neonatal hypoxia-ischemia
KR102026142B1 (en) Anti-Cancer and Anti-Metastasis Composition Comprising CRIF1 Antagonist
WO2017160797A1 (en) Combination therapy with c-myc nucleic acid inhibitors and selective cdk7 inhibitors
US20230151363A1 (en) Modified short-interfering rna compositions and their use in the treatment of cancer
KR20240050416A (en) Pharmaceutical composition for inhibiting cancer invasion and method for inhibiting cancer invasion
CN117881404A (en) Pharmaceutical composition for inhibiting infiltration of cancer and method for inhibiting infiltration of cancer

Legal Events

Date Code Title Description
E701 Decision to grant or registration of patent right
GRNT Written decision to grant